Summary:
The Coronavirus Disease 2019 (COVID-19) pandemic has presented a major threat to public health worldwide alongside unprecedented global economic and social implications. In the absence of a “gold standard” treatment, the rapid development of a safe and effective vaccine is considered the most promising way to control the pandemic. In recent years, traditional vaccine technologies have seemed insufficient to provide global protection against the rapid spread of emerging pandemics. Therefore, the establishment of novel approaches that are independent of whole pathogen cultivation, cost-effective, and able to be rapidly developed and produced on a large scale are of paramount importance for global health. This article summarizes the current efforts to develop a COVID-19 vaccine, including the ongoing and future anticipated clinical trials. We also provide plastic and reconstructive surgeons with insight into the novel technologies currently utilized for COVID-19 vaccine development, focusing on the very promising viral-vector-based and gene-based vaccine technologies. Each platform has its own advantages and disadvantages related to its efficacy and ability to induce certain immune responses, manufacturing capacity, and safety for human use. Once the fundamental key challenges have been addressed for viral-vector-based and gene-based vaccines, these novel technologies may become helpful in winning the fight against COVID-19 and transforming the future of health care.
Science knows no country, because knowledge belongs to humanity, and is the torch which illuminates the world.
—Louis Pasteur
INTRODUCTION
The Coronavirus Disease 2019 (COVID-19) pandemic has presented a major threat to public health worldwide alongside unprecedented global economic and social implications. Identified in Wuhan, Hubei Province, China, in December 2019, the novel coronavirus, Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2),1 has infected a total of 20,840,381 individuals, causing 754,566 global deaths as of August 14, 2020. An estimated 5,248,722 cases have been confirmed in the United States, and the death toll there has surpassed 160,000.2
COVID-19 has a relatively high degree of pre-symptomatic transmission,3,4 high mortality rate, severe morbidity rate, and relatively long hospitalization period.5 As a result, the fight against COVID-19 has required extensive interventions such as quarantine, social distancing, isolation of infected populations, border closures, school shutdowns, and extensive lockdowns to contain the virus, “flatten the curve,” and save lives.6 The COVID-19 pandemic has changed the landscape of plastic surgery,7 restructured the daily experience of plastic surgery practices,7–11 altered the way residents and fellows are trained,12,13 substantially impacted research,14,15 and revolutionized patient care. The implications of the current COVID-19 pandemic on plastic surgery are summarized in Table 1.
Table 1.
Effects of COVID-19 on Plastic Surgery
Aspect Affected | Effects |
---|---|
Clinical care | • Rescheduling/cancellation of surgeries, procedures, and in-person appointments |
• Practice closures | |
• Telehealth for preoperative and postoperative discussions, no in-person physical examination | |
• Implementation of patient flow plans that allow for social distancing protocols | |
• Reassessment of cleaning and disinfecting protocols | |
• PPE requirements for surgeons, anesthesia, and staff | |
• Patients’ screening upon entrance, “no visitors” policy, masking requirement | |
• Updated safety protocols for elective surgery | |
• New informed consent form for COVID-19 risk | |
• Relatively bear market for elective surgery | |
Education and training | Residency and Fellowship Training Programs• Redeployment to ICUs and emergency rooms |
• Utilization of virtual platforms for didactic sessions: daily team briefings, morning conferences, and grand rounds and nationally integrated didactics | |
• Case category minimum requirements by the ACGME and the ABPS might not be reached due to decreased surgical volumes | |
Plastic Surgery Residency Application Cycle• Cancellation of away rotations | |
• Virtual away rotations and program-applicant communication via social media | |
• Utilization of virtual platforms for didactic sessions | |
• Online residency interviews and virtual visits | |
• Rescheduling/cancellation of USMLE, subsequent changes to the ERAS application cycle and adjusted deadlines | |
• Postponement and cancellation of national and regional conferences | |
Research | • Laboratory closures |
• Suspension of clinical trials | |
• Postponement and cancellation of national and regional conferences | |
• Utilization of virtual platforms for research and didactic sessions |
ABPS, American Board of Plastic Surgery; ACGME, Accreditation Council for Graduate Medical Education; COVID-19, Coronavirus disease 2019; ERAS, Electronic Residency Application Service; ICU, intensive care unit; PPE, personal protective equipment; USMLE, United States Medical Licensing Examination.
Due to the profound global implications of the COVID-19 crisis, there has been an unprecedented race to develop treatments and vaccines against SARS-CoV-2. Multiple clinical trials are underway to define potential roles for antiviral agents and specific immunomodulators16–18 as well as passive immunization with convalescent plasma.19 However, no “gold standard” treatment or prophylactic medication has been approved for COVID-19. Optimizing supportive care for COVID-19 positive patients remains the mainstay of therapy, including oxygen, mechanical ventilation, and treatment of the sequelae and complications.20 With multiple waves of illness anticipated,21 and in the absence of approved treatments, the development of a safe and effective vaccine will be a game-changing step in the global fight against COVID-19 and is considered the most promising way to eradicate the virus.22
This article summarizes the current efforts to develop a SARS-CoV-2 vaccine, including the ongoing and future anticipated clinical trials. We also provide plastic and reconstructive surgeons with insight into the novel technologies currently utilized for SARS-CoV-2 vaccine development, focusing on the very promising viral-vector-based and gene-based vaccine technologies (Fig. 1).
Fig. 1.
Pie charts showing the development of COVID-19 candidate vaccines: (A) clinical trials and (B) preclinical trials.
VACCINES: IMMUNOLOGICAL PRINCIPLES AND VACCINE DEVELOPMENT STRATEGIES
Since the development of the first vaccine by Dr. Edward Jenner more than 200 years ago,23 vaccinations have made an enormous contribution to global health.24 However, the development process for conventional vaccines takes more than 10 years25 and requires 4 Phases,26–28 as summarized in Figure 2. Therefore, the lack of time remains a major barrier for safe and effective vaccine development in outbreak situations.
Fig. 2.
Diagram displaying the various phases of a clinical trial.
Immunological Principles of Vaccination
Immunization can be derived from either passive or active immunization.29 Passive immunization occurs with the transfer or administration of already preformed antibodies, providing temporary immunity.29 One investigational treatment being explored for COVID-19 is the use of convalescent plasma collected from previously infected individuals, which is administered via direct transfusion to COVID-19 patients, in an attempt to confer passive immunity.30–34
Active immunization occurs with the exposure to an antigen and typically produces long-term immunity due to the immune system stimulation.29 The immune system is divided into two main subsystems: the innate system, which provides an initial, non-specific response, with no memory induced, and the adaptive system, which provides a later, antigen-specific response and induces immunological memory.35 When antigens are introduced into the bloodstream via infection or vaccination, they are captured and processed by Antigen Presenting Cells (APCs), which then display an antigen-derived peptide fragment on their surfaces. APCs then migrate to lymph nodes, and activate T helper cells through a process called “antigenic presentation.” T helper cells stimulate both arms of the adaptive immunity: humoral (antibody-based) and cellular.
Humoral immunity is achieved via differentiation, proliferation, and maturation of B-lymphocytes into antibody-secreting plasma cells and memory lymphocytes of the same antigenic specificity, and cell-mediated immunity is achieved via activation of naive cytotoxic T cells to active, antigen-specific cells.29 The stimulation of both the humoral and cell-mediated arms of the adaptive system by the production of effector cells (plasma B cells for humoral immunity and active cytotoxic T cells for cell-medicated immunity) and memory cells is required to ensure protection upon re-exposure to the same antigen.35 Figure 3 illustrates the immune stimulation response in detail.
Fig. 3.
Illustration depicting the stimulation of humoral and cellular immune responses.
Conventional Vaccine Platforms
The main conventional vaccine technologies include inactivated vaccines (which utilize a killed pathogen), subunit/recombinant vaccines (which utilize specific pieces of the pathogen administered along with adjuvants), toxoid vaccines (pathogen-toxin-based), and live attenuated vaccines.36 Each platform has its own advantages and disadvantages related to its efficacy, immunogenicity, and ability to induce certain immune responses, manufacturing capacity, and safety for human use, as summarized in Table 2.36–42
Table 2.
SARS-CoV-2 Vaccine Development Platforms
Platform (Refs) |
No. Clinical Trials | No. Pre-clinical Studies | Platform Status | Advantages | Disadvantages | Existing Licensed Vaccines |
---|---|---|---|---|---|---|
Inactivated | 5 | 9 | Licensed | Safety: • Cannot replicate |
Safety: • Infection risk |
HAV, influenza (shot only), Polio (shot only), rabies |
• No adjuvants required | ||||||
Efficacy: • High potency • Multivalent |
Efficacy: • Often induces weaker immune responses than other methods |
|||||
Production and Manufacturing: • Complex manufacturing—lengthy, relatively small quantities produced | ||||||
• Requires vigorous quality control | ||||||
Live attenuated virus | 0 | 3 | Licensed | Safety: • No adjuvants required |
Safety: • Reversion risk |
Measles, mumps, rubella (MMR combined vaccine), rotavirus, smallpox, chickenpox, yellow fever |
• Inactivation may lead to undesired effects and exacerbated disease | ||||||
• Adverse effects in immune-compromised | ||||||
Efficacy: • High potency • Multivalent • Induction of long-lived responses |
Efficacy: • Inactivation may hamper protective immune responses |
|||||
Production and Manufacturing: • Requires whole pathogen cultivation, high biosafety level, and specialized laboratories | ||||||
• Relatively high production costs requires dedicated production processes and facilities for each vaccine | ||||||
• Insufficient production capacities for global vaccination | ||||||
Viral vector | Non-replicating: 5 Replicating: 1 |
Non- replicating: 19 Replicating: 17 |
Experimental | Safety: • Favorable safety profile—whole pathogen cultivation not required • No adjuvants required |
Safety: • Potential induction of anti-vector immunity |
Non-replicating: none Replicating: dengue fever |
• Potential environmental risks associated with the release of genetically modified organisms | ||||||
• Potential integration into the host genome and persistent replication of attenuated vaccines | ||||||
• Potential risk for infection | ||||||
Efficacy: • Previous infection/pre-existing immunity to the viral vector may hamper immune responses | ||||||
• Inability to administer multiple times | ||||||
Production and Manufacturing: • Cell-based and antigen-dependent manufacturing | ||||||
• Requires vigorous monitoring | ||||||
• Complex manufacturing | ||||||
Efficacy: • Able to induce potent, antigen-specific cellular and humoral immune responses |
||||||
• Strong innate immune response | ||||||
Production and manufacturing: • High producibility—independent of whole pathogen cultivation | ||||||
• High specificity and accuracy—can be engineered easily to accurately express any antigen of choice, specific targeting, and processing in the cell due to antigen delivery as genetic information | ||||||
• High versatility—allows large insertions in genome and therefore the development of a large variety of vaccines | ||||||
• Sufficient production capacities for global vaccination due to established high yield production processes with means of upscaling | ||||||
Protein Subunit | 7 | 50 | Licensed | Safety: • Non-infectious |
Safety: • Requires additional adjuvants |
Hib, HBV, HPV, Whooping cough (part of the DTaP combined vaccine), Pneumococcal disease, Meningococcal disease, Shingles |
Efficacy: • Strong humoral response |
Efficacy: • Multiple doses are usually required |
|||||
Production and Manufacturing: • Requires dedicated production processes, facilities and stability assays for each vaccine | ||||||
• Multivalent formulation can be challenging | ||||||
RNA | 6 | 16 | Experimental | Safety: • Favorable safety profile—whole pathogen cultivation not required, non-infectious |
Safety: • Potential risk for severe adverse reactions |
None |
• No interaction with the host-cell DNA, avoiding the potential risk of genomic integration | ||||||
• Natural degradation and lack of persistence in cells | ||||||
Efficacy: • Generation of potent humoral and cellular immune responses | ||||||
• Very potent innate immune response | ||||||
• Can be administered multiple times (boosting) | ||||||
Administration: • Can be administered by different routes, do not require additional administration devices | ||||||
Production and Manufacturing: • High specificity—able to encode any antigen of choice |
• Production and Manufacturing: • Relatively high production costs |
|||||
• High versatility—able to produce different vaccines using the same established production process and facility. | ||||||
• Safe, rapid, and scalable production—based on in vitro systems that are simple to monitor, production free of animal-derived products | ||||||
• Small amounts of expressed protein required due to amplification by the immune system | ||||||
DNA | 4 | 12 | Experimental | Safety: • Favorable safety profile— whole pathogen cultivation not required, non-infectious |
Safety: • Potential long-term persistence, risk of genomic integration, potentially leading to mutagenesis and oncogenesis. |
None |
• Potential generation of autoantibodies | ||||||
• Potential adverse effects due to cytokines/co-stimulatory molecules expression used to enhance DNA immunogenicity | ||||||
Efficacy: • Generation of potent humoral and cellular immune responses |
Efficacy: • Low immunogenicity in humans |
|||||
• High stability | ||||||
Production and Manufacturing: • High specificity—able to encode any antigen of choice |
Production and Manufacturing: • Requires additional methods to enhance DNA uptake, expression, and immunogenicity: delivery devices such as gene gun, needle, jet injection, and in vivo electroporation and molecular adjuvants |
|||||
• Safe, rapid and scalable production—based on in vitro systems that are simple to monitor, production free of animal-derived products | ||||||
Other platforms | 1 | 12 | N/A | N/A | N/A | N/A |
DNA, Deoxyribonucleic acid; DTaP, diphtheria, tetanus, and acellular pertussis; HAV, Hepatitis A virus; Hib, Haemophilus influenzae type b; HBV, Hepatitis B virus; HPV, human papillomavirus; RNA, ribonucleic acid.
Novel Vaccine Platforms
In recent years, traditional vaccine technologies that consist of working with a virulent pathogen during manufacturing seem insufficient to provide global protection against the spread of emerging pandemics. Therefore, the establishment of novel approaches that are independent of pathogen cultivation (a relatively lengthy and high-risk process), are cost-effective, and could be rapidly developed and produced on a large scale, are of paramount importance for global health.41
Two promising novel platforms that have generated significant attention in recent years due to their potential use for a variety of applications include the viral-vector-based and gene-based vaccine technologies. Their advantages over conventional approaches highlight the role of these platforms in the new era of vaccinology, as potential game-changers in epidemics and emerging diseases.43 The strengths and weaknesses of these platforms are summarized in Table 2 and discussed below.
Viral-vector-based Vaccines
Viral vector vaccines use separate, genetically engineered, viruses to carry DNA of target antigens into human cells. The DNA contained in the viral vector encodes antigens that, once expressed in the infected human cells, elicit antigen-specific humoral and cell-mediated immune responses via antigen presentation (Fig. 3).44 A variety of viruses have been employed as viral vectors,45 with Adenovirus (Ad) vectors being the most commonly utilized due to their various advantages. Ad vectors’ advantages over other viral vectors include their ability to enter a broad range of target cells in humans,46 deliver various target antigens and large DNA insertions,47 and induce potent humoral and cellular responses.48 However, utilizing an unrelated virus for delivery poses several challenges in terms of manufacturing, safety, and immunogenicity, as summarized in Table 2.
Gene-based Vaccines
Gene-based vaccine platforms consist of genetic sequences in the form of plasmid DNA49 and mRNA.50 Once injected intramuscularly (IM), the genetic sequence enters the myocytes51 to achieve encoding of the desired antigen.52 The endogenously synthesized antigens are then secreted from myocytes for cross presentation and consecutive stimulation of humoral and cellular responses, as illustrated in Figure 3.53 The main advantages of the gene-based platform include the construction of antigens directly from the genetic sequence of the desired protein and a generic manufacturing process, which allows for efficient production of different vaccines using the same established processes and facilities, given the genetic sequence is available.54
Unlike conventional vaccine technologies, which consist of a lengthy process of inactivating or attenuating a live pathogen or making a recombinant protein, making a gene-based vaccine is relatively rapid and potentially low-risk, eliminating the need to work with a virulent pathogen during manufacturing.38 In addition, plasmid DNA and mRNA vaccine constructs encode only the antigen of interest, avoiding other redundant, potentially detrimental proteins and the replication of infectious viral particles.55
However, since their initial presentation in 1990,56 there has never been a commercial vaccine utilizing gene-based technologies approved for use. There have been many technical challenges to overcome to enable the promise of the gene-based platform,57 including optimizing the delivery of these foreign nucleic acids into human cells58,59 and increasing the potency, stability, and expression of the encoded protein.38,55 These goals have been achieved by new formulations, including lipids, polymers, and novel delivery devices for improved intracellular delivery and stability, and strong molecular adjuvants for improved potency.60,61
DNA-based Vaccines consist of antigen-encoding plasmids. While allowing a relatively simple, safe, and time-saving production process, the DNA-based platform poses several challenges related to administration, safety, and immunogenicity, as summarized in Table 2. Unlike DNA vaccines, mRNA-based vaccines translate directly into the cytoplasm, undergo natural degradation,50 and cannot integrate into the host genome.41 These advantages in terms of safety, efficacy, and manufacturing make the mRNA technology a promising avenue for a rapid response to the emerging COVID-19 pandemic, and are summarized, alongside the disadvantages, in Table 2.
COVID-19 Vaccine Development: Current Clinical Trials
Finding the most suitable target site for SARS-COV-2 vaccine development is extremely important. The SARS-CoV-2 coronavirus belongs to the subfamily of Coronavirinae, with a genomic structure of (+) ss-RNA.62 Hoffman et al.63 described the SARS-CoV-2 cell entry and replication mechanisms in detail and Bouhaddou et al.64 recently demonstrated the role of virus-containing filopodia induced in host cells by SARS-CoV-2 in viral replication. Cell entry of SARS-CoV-2 is orchestrated by the viral spike (S) proteins, which give the virus its characteristic corona-like morphology, via binding the host cellular receptors.63,65 Due to its pivotal role, a vaccine against S protein can prevent SARS-CoV-2’s proliferation and spread.66
As of August 14, 2020, there have been 29 vaccine candidates in clinical trials, as summarized in Table 3 and Figure 1. In total, 13 trials are currently in Phase 1, 8 in joint Phase 1/2, 2 in Phase 2 and 6 in Phase 3. An estimated 138 vaccine candidates are currently in preclinical studies.67 While 12 clinical trials are based on traditional techniques utilizing inactivated viruses (5 trials),68–73 and protein subunits (7 trials),74–77 16 trials utilize novel platforms including the viral vector-based platform (6 trials),78–84 and gene-based platform (10 trials: 6 mRNA-based, 4 DNA-based).85–94 Current viral-vector-based and gene-based vaccines under development target the S protein of SARS-CoV-2.86,88
Table 3.
Candidate Vaccines against SARS-CoV-2 Currently in Clinical Trials
S. No. | Vaccine Name | Developer (Country) | Platform | Phase 1 Start Date (mm/dd/yyyy) | Current Phase | Participants’ Age and Sample Size (N)* | Mode of Administration | Vaccination Schedule* |
Same Platform for Other Viral Candidates |
---|---|---|---|---|---|---|---|---|---|
1 | N/A | Beijing Institute of Biological Products and Sinopharm (China) | Inactivated | 04/28/2020 | Phase 3 (ChiCTR2000034780) Phase 1/2 (ChiCTR2000032459) |
• Ages: 18 and over • N = 15,000 |
IM | Day 0, 14 or 0, 21 | |
2 | N/A | Wuhan Institute of Biological Products and Sinopharm (China) | Inactivated | 04/11/2020 | Phase 3 (ChiCTR2000034780) Phase 1/2 (ChiCTR2000031809) |
• Ages: 18 and over • N = 15,000 |
IM | Day 0, 14 or 0, 21 | |
3 | PiCoVacc/ PROFISCOV | Sinovac (China) | Inactivated | 04/16/2020 | Phase 3 (NCT04456595) Phase 1/2 (NCT04383574) (NCT04352608) |
• Ages: 18 and over • N = 8870 |
IM | Day 0, 14 | SARS |
4 | N/A | Institute of Medical Biology, Chinese Academy of Medical Sciences (China) | Inactivated | 05/15/2020 | Phase 1/2 (NCT04470609) Phase 1 (NCT04412538) |
• Ages: 18–59, 60 and over • N = 942, 471 |
IM | Day 0, 28 | |
5 | BBV152 | Bharat Biotech (India) | Inactivated | 07/13/2020 | Phase 1/2 (NCT04471519) |
• Ages: 16–65 • N = 1125 |
IM | Day 0, 14 | |
6 | ChAdOx1 nCoV-19 (AZD1222) | University of Oxford and AstraZeneca (UK) | Viral vector (non- replicating) | 03/19/2020 | Phase 3 (ISRCTN89951424) Phase 2b/3 (2020-001228-32) Phase 1/2 (2020-001072-15) |
• Ages: 18–55 • N = 10,260; 30,000; 2000 |
IM | Day 0 | MERS, influenza, TB, Chikungunya, Zika, MenB, plague |
7 | Adenovirus Type 5 Vector (Ad5-nCoV) |
CanSino Biological and Beijing Institute of Biotechnology (China) | Viral vector (non- replicating) | 03/16/2020 | Phase 2 (ChiCTR2000031781) Phase 1 (ChiCTR2000030906) |
• Ages: 18 and over • N = 500 |
IM | Day 0 | EBOV |
8 | Gam-COVID-Vac | Gamaleya Research Institute (Russia) | Viral vector (non-replicating) | 06/17/2020 | Phase 1 (NCT04436471) (NCT04437875) |
• Ages: 18–60 • N = 38 |
IM | Day 0 | |
9 | Ad26COVS1 | Janssen Pharmaceutical Companies (USA, Belgium) | Viral vector (non- replicating) | 07/15/2020 | Phase 1/2 (NCT04436276) |
• Ages: 18-55 • N = 1045 |
IM | Day 0, 56 | |
10 | N/A | ReiThera/LEUKOCARE/Univercells (Italy, Germany, Belgium) | Viral vector (non- replicating) | N/A | Phase 1 )2020-002835-31( |
N/A | IM | N/A | |
11 | COVID-19-101 | Institute Pasteur/Themis/Univ. of Pittsburg CVR/Merck Sharp & Dohme (Belgium, France) | Viral vector (replicating) | 08/10/2020 | Phase 1 (NCT04497298) |
• Ages: 18–55 • N = 90 |
IM | Day 0, 28 | |
12 | NVX-CoV2373 | Novavax (USA, Australia) | Protein subunit | 05/25/2020 | Phase 1/2 (NCT04368988) |
• Ages: 18–59 • N = 131 |
IM | Day 0, 21 | RSV; CCHF, HPV, VZV, EBOV |
13 | SCB-201 | Clover Biopharmaceuticals Inc./GSK/Dynavax (Australia) | Protein subunit | 06/19/2020 | Phase 1 (NCT04405908) |
• Ages: 18–75 | IM | Day 0, 21 | HIV, REV Influenza |
14 | N/A | Anhui Zhifei Longcom Biopharmaceutical/ Institute of Microbiology, Chinese Academy of Sciences (China) | Protein subunit | 06/22/2020 | Phase 2 (NCT04466085) Phase 1 (NCT04445194) |
• Ages: 18–59 • N = 900 |
IM | Day 0, 28 or 0, 28, 56 | MERS |
15 | COVAX19 | Vaxine Pty Ltd/Medytox (Australia) | Protein subunit | 06/30/2020 | Phase 1 (NCT04453852) |
• Ages: 18–65 • N = 40 |
IM | N/A | |
16 | KBP 201 | Kentucky Bioprocessing, Inc (USA) | Protein subunit | 09/14/2020 | Phase 1/2 (NCT04453852) |
• Ages: 18–70 • N = 180 |
IM | Day 0, 21 | |
17 | N/A | University of Queensland/CSL/Seqirus (Australia) | Protein subunit | 07/13/2020 | Phase 1 (ACTRN12620000674932p) |
• Ages: 18–55 • N = 120 |
IM | Day 0, 28 | |
18 | MVC-COV1901 | Medigen Vaccine Biologics Corporation/NIAID/Dynavax (Taiwan) | Protein subunit | 09/01/2020 | Phase 1 (NCT04487210) |
• Ages: 20–50 • N = 45 |
IM | Day 0, 28 | |
19 | mRNA-1273 | Moderna and NIAID (USA) | mRNA | 03/16/2020 | Phase 3 (NCT04470427) Phase 2 (NCT04405076) Phase 1 (NCT04283461) |
• Ages: 18 and over • N = 30,000 |
IM | Day 0, 28 | Multiple candidates |
20 | BNT162 | BioNTech and Pfizer (Germany and USA) | mRNA | 04/29/2020 | Phase 3 (NCT04368728) Phase 1/2 (2020–001038–36) (NCT04368728) |
• Ages: 18–85 • N = 29,481 |
IM | Day 0, 28 | |
21 | COVAC1 | Imperial College London (UK) | mRNA | 04/01/2020 | Phase 1 (ISRCTN17072692) |
• Ages: 18–45 • N = 320 |
IM | N/A | EBOV; LASV, MARV, Inf (H7N9), Rabies |
22 | CVnCoV | Curevac (Germany, Belgium) | mRNA | 06/18/2020 | Phase 1 (NCT04449276) |
• Ages: 18–60 • N = 168 |
IM | Day 0, 28 | Rabies, LASV, YFV; MERS, InfA, Zika, dengue, NPV |
23 | N/A | People’s Liberation Army (PLA) Academy of Military Sciences/Walvax Biotech (China) | mRNA | 06/25/2020 | Phase 1 (ChiCTR2000034112) |
• Ages: 18–80 • N = 168 |
IM | N/A | |
24 | ARCT-021 | Arcturus/Duke-NUS (Singapore) | mRNA | 08/10/2020 | Phase 1/2 (NCT04480957) |
• Ages: 21–80 • N = 92 |
IM | Day 0,14 or 0,28 | |
25 | INO-4800 | Inovio Pharmaceuticals (USA) | DNA | 04/03/2020 | Phase 1/2 (NCT04336410) |
• Ages: 18 and over • N = 120 |
ID | Day 0, 28 | Multiple candidates |
26 | nCov | Cadila Healthcare Limited (India) | DNA | 07/04/2020 | Phase 1/2 (CTRI/2020/07/026352) |
• Ages: 18–55 • N = 1084 |
ID | Day 0, 28, 56 | |
27 | GX-19 | Genexine Consortium (Korea) | DNA | 06/17/2020 | Phase 1 (NCT04445389) |
• Ages: 18–50 • N = 190 |
IM | Day 0, 28 | |
28 | N/A | Osaka University/AnGes/Takara Bio (Japan) | DNA | N/A | Phase 1 (NCT04463472) |
• Ages: 20–65 • N = 30 |
IM | Day 0, 14 | |
29 | N/A | Medicago Inc. (Quebec, Canada) | VLP | 07/10/2020 | Phase 1 (NCT04450004) |
• Ages: 18–55 • N = 180 |
IM | Day 0, 21 |
*Data reflect the most advanced clinical phase available.
No, number; N/A, not available; CCHF, Crimean-Congo Hemorrhagic Fever; DNA, Deoxyribonucleic acid; EBOV, Ebola virus; GMFR, Geometric mean fold rise; GMI, Geometric Mean Increase; GMR, Geometric Mean Ratio; GMT, Geometric Mean Titer; HPV, Human Papillomavirus; IFN-γ, Interferon gamma; IgG, Immunoglobulin G; IgM, Immunoglobulin M; LASV, Lassa virus; LNP, Lipid nanoparticles; MARV, Marburg virus; Inf, influenza; MenB, Serogroup B Meningococcal; MERS, Middle East Respiratory Syndrome; mRNA, messenger ribonucleic acid; NIAID, National Institute of Allergy and Infectious Diseases; NPV, nuclear polyhedrosis virus; RNA, Ribonucleic acid; RSV, Respiratory syncytial virus; SARS-CoV-2, Severe acute respiratory syndrome coronavirus 2; TB, Tuberculosis; VZV, Varicella-zoster virus; YFV, yellow fever virus.
Viral-vector-based Clinical Trials
Three viral-vector-based vaccines are currently under clinical trials (Table 3). The Adenovirus Type 5 Vector (Ad5-nCoV) and ChAdOx1 nCoV-19 trials are the furthest along in development and will be discussed below.
Adenoviral Vectors of Human Origin: Ad5-nCoV
Ad5-nCoV is a recombinant adenovirus type-5 (Ad5) vectored COVID-19 vaccine expressing the S protein of SARS-CoV-2.95 Ad5 is a non-replicating vector of human origin, and one of the most widely used adenoviral vectors.96 However, the widespread pre-existing immunity to Ad5 among the human population might hinder its immunogenicity and hamper its clinical use.97 Phase 1 results have recently been published,80,95 demonstrating that the Ad5 vectored COVID-19 vaccine was tolerable and immunogenic at 28 days post-vaccination. Most adverse reactions were mild or moderate in severity, with the most common adverse reactions being fever, fatigue, headache, and muscle pain. No serious adverse events were noted within 28 days post-vaccination. Neutralizing antibodies increased significantly at day 14, and peaked 28 days post-vaccination, while specific T-cell response peaked at day 14 after one administration of the vaccine. However, in patients with pre-existing anti-Ad5 immunity, both the specific antibody response and T-cell response induced by vaccination were diminished.95 The recently published Phase II results81,98 have confirmed Phase I results, demonstrating that the Ad5-vectored COVID-19 vaccine was safe, and induced significant immune responses in the majority of recipients after a single immunization. Phase III results are expected before the end of 2020.
Adenoviral Vectors of Non-human Origin: ChAdOx1 nCoV-19 (AZD1222)
Adenoviral vectors of non-human origin induce enhanced memory and more poly-functional CD8+ T cells compared with Ad5 and are less likely to be hampered by pre-existing immunity.48,99 ChAdOx1 nCoV-19 is a replication-deficient chimpanzee adenovirus expressing the S protein of SARS-CoV-2. Phase I/II results have recently been published,79,100 demonstrating an acceptable safety profile of ChAdOx1 nCoV-19 with no serious adverse events. The vaccine induced both humoral and cellular immune responses and all participants had neutralizing activity after a booster dose. Phase III is currently ongoing in the UK, the USA, Brazil, and South Africa, and results are expected in the Fall of 2020.83,101
Gene-based Vaccines Clinical Trials
Totally, 4 DNA-based and 5 mRNA-based vaccines are currently in clinical trials (see Table 3). The m-RNA-1273 trial is the furthest along in development and will be discussed below.
m-RNA-1273
mRNA-1273 developed in collaboration with the National Institute of Allergy and Infectious Disease (NIAID) of the National Institute of Health (NIH) is considered one of the frontrunners in vaccine development. The mRNA-1273 vaccine consists of mRNA of the S protein synthesized in vitro, and coated with lipid nanoparticles for effective delivery.
Phase I results have recently been published,86,102 demonstrating that mRNA-1273 was generally safe and well tolerated, with 1 incidence of a grade 3 adverse event (erythema around the injection site), and 3 incidences of grade 3 systemic symptoms (including fever, muscle pain and headache) seen at the highest dose group, only following the 2nd dose. All adverse events have been transient and self-resolving. No grade 4 adverse events or serious adverse events have been reported. Phase II is currently ongoing,85 and Phase III is expected to commence before the end of July 2020 in collaboration with the NIAID. Phase III results are expected before the end of 2020.102
Looming Challenges of COVID-19 Vaccine Development
While the preliminary results of the discussed trials are encouraging, whether a vaccine generates the needed types of immune responses to result in protective efficacy is unknown and cannot be predicted by Phase I studies. There still remain significant questions and uncertainties as to clinical efficacy and effects of potential mutations on the vaccine’s immunogenicity and long-term efficacy, safety, and potential benefit for specific target populations.
Clinical Efficacy and Long-term Immunity
One of the main questions that arise is whether a COVID-19 vaccine would be able to provide immunity, when it is still uncertain whether previously infected patients are protected from reinfection.103–107 Research in other coronavirus species has shown that immunity may not be long-lasting, with 2–3 years of protection estimated from work with SARS and MERS.102,108,109
To date, there is no evidence for cases of SARS-CoV-2 reinfections, 103 and preliminary evidence in humans110 and rhesus macaques111 suggests that acquired immunity may protect from future reinfection, at least temporarily104,111–113 with most patients who recovered from COVID-19 producing sufficient amounts of neutralizing antibodies to protect against reinfection. A recently reported case series also indicated sufficient neutralizing antibody titers in convalescent plasma to neutralize SARS-CoV-2 in 5 COVID-19 patients, who all recovered after treatment.114 While further research is needed to assess the benefits and risks of convalescent plasma therapy for people with COVID-19,33,34 these preliminary findings provide hope for the development of an effective vaccine against SARS-CoV-2.32 However, there have been few reports of post-recovery positive PCR tests performed in asymptomatic or mildly symptomatic patients, which could be explained as either false-negative results, prolonged viral clearance, and shedding or reinfection.106,107,115–117
Further points that need to be addressed to better understand the immune responses to SARS-CoV-2 and the optimal vaccine profile and administration regimens include the lack of correlation between antibody titer rates and clinical improvement, the durability of neutralizing antibodies, and their correlation with durable immunity.118,119 The current gaps in knowledge highlight the importance of inducing potent humoral and cellular immune responses as potentially generated by viral-vector-based and gene-based vaccines.
Potential mutations in the S protein may also affect the long-term efficacy of a vaccine. There has been direct evidence of functionally meaningful S protein mutations that appear to mediate a higher binding affinity when compared with previous SARS viruses.120 Therefore, it is difficult to ensure that the current novel vaccines targeting SARS-CoV-2 protein S could be used for a long term. This highlights the importance of a cost-effective platform that is able to produce different vaccines rapidly and safely using existing production processes and already established manufacturing infrastructure. This is one of the main potential strengths of the gene-based technology over the conventional vaccine development platforms.
Clinical Safety and Adverse Events
There have been previous reports of systemic reactions to mRNA, DNA, and viral-vectored vaccines, including adverse reactions identified in the Ad5-nCoV95 and mRNA-1273 Phase 1 trials.89 These outcomes have raised concerns about the safety of these novel platforms. As previously discussed, one of the main concerns of utilizing mRNA-based platforms is the potential toxicity of synthetically formulated mRNA due to its inherent inflammatory nature.55 The use of DNA-based vaccines and viral-vector-based vaccines raises safety concerns due to their potential long-term persistence,121,122 genome integration,123,124 autoantibody generation, potential induction of anti-vector immunity,124 and adverse effects due to co-stimulatory molecule expression.125
These safety concerns cannot be fully investigated by pre-clinical studies because humans may respond differently than the animal models used in the pre-clinical safety testing.126 Therefore, it is of vital importance to fully characterize the potential risks of these novel platforms and adjust dosing schemes accordingly. This holds true especially for mRNA-based vaccines, where repeated administration (boosting) is needed for generating the desired neutralizing antibody titer levels.127
Clinical Benefit for Specific Target Populations
A major question that requires further investigation is whether the elderly and immunocompromised populations, who experience higher clinical attack rates and a more severe clinical course,128 would be able to mount a sufficiently robust antibody response to provide immunity in response to the vaccine. Most current trials are designed to include healthy elderly participants as part of advanced clinical phases; however, safety and potential efficacy should be established to include immunocompromised patients.
CONCLUSIONS
In conclusion, the rapid development of an effective and safe vaccine has become the most promising way to control the COVID-19 pandemic. Over 20 candidate vaccines are in clinical trials and over 100 are in preclinical trials, utilizing both conventional and novel technologies. The viral-vector-based and gene-based vaccine technologies are promising novel platforms that have generated significant attention in recent years due to their potential use for a variety of applications. Their various advantages over conventional approaches highlight their role in the new era of vaccinology as potential game-changers in epidemics and emerging diseases. Once the fundamental key challenges have been addressed for viral-vector-based and gene-based vaccines, these novel technologies may become helpful in winning the fight against COVID-19 and in transforming the future of health care.
In the words of Louis Pasteur: In the field of observation, chance favors only the prepared mind.
Footnotes
Published online 15 October 2020.
Disclosure: None of the authors has a financial interest in any of the products, devices, or drugs mentioned in this article.
REFERENCES
- 1.Wang C, Horby PW, Hayden FG, et al. A novel coronavirus outbreak of global health concern. Lancet. 2020;395:470–473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Coronavirus Research Center. COVID-19 Dashboard by The Center for Systems Science and Engineering (CSSE) at Johns Hopkins University. Available at https://coronavirus.jhu.edu/map.html. Accessed June 11, 2020.
- 3.Gandhi M, Yokoe DS, Havlir DV. Asymptomatic transmission, the Achilles’ heel of current strategies to control COVID-19. N Engl J Med. 2020;382:2158–2160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Arons MM, Hatfield KM, Reddy SC, et al. ; Public Health–Seattle and King County and CDC COVID-19 Investigation Team. Presymptomatic SARS-CoV-2 infections and transmission in a skilled nursing facility. N Engl J Med. 2020;382:2081–2090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Squitieri L, Chung KC. Surviving the COVID-19 pandemic: Surge capacity planning for nonemergent surgery. Plast Reconstr Surg. 2020;146:437–446. [DOI] [PubMed] [Google Scholar]
- 6.Anderson RM, Heesterbeek H, Klinkenberg D, et al. How will country-based mitigation measures influence the course of the COVID-19 epidemic? Lancet. 2020;395:931–934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Rohrich RJ, Hamilton KL, Avashia Y, et al. The COVID-19 pandemic: changing lives and lessons learned. Plast Reconstr Surg Glob Open. 2020;8:e2854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Hollander JE, Carr BG. Virtually perfect? telemedicine for COVID-19. N Engl J Med. 2020;382:1679–1681. [DOI] [PubMed] [Google Scholar]
- 9.Shokri T, Lighthall JG. Telemedicine in the era of the COVID-19 pandemic: Implications in facial plastic surgery. Facial Plast Surg Aesthet Med. 2020;22:155–156. [DOI] [PubMed] [Google Scholar]
- 10.Sarac BA, Schoenbrunner AR, Wilson SC, et al. Coronavirus disease 2019 state guidelines on elective surgery: considerations for plastic and reconstructive surgeons. Plast Reconstr Surg Glob Open. 2020;8:e2904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Schoenbrunner AR, Sarac BA, Janis JE. A summary of recommendations for plastic surgeons during the coronavirus disease 2019 outbreak. Plast Reconstr Surg Glob Open. 2020;8:e3039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Taub PJ. Plastic surgeons in the time of a pandemic: thoughts from the front line. Plast Reconstr Surg. 2020;146:458–459. [DOI] [PubMed] [Google Scholar]
- 13.Cho DY, Yu JL, Um GT, et al. The early effects of COVID-19 on plastic surgery residency training: the University of Washington experience. Plast Reconstr Surg. 2020;146:447–454. [DOI] [PubMed] [Google Scholar]
- 14.FDA Guidance on Conduct of Clinical Trials of Medical Products during COVID-19 Public Health Emergency. Available at https://www.fda.gov/media/136238/download. Accessed June 25, 2020.
- 15.Asaad M, Habibullah NK, Butler CE. The impact of COVID-19 on clinical trials. Ann Surg. 2020;272:e222–e223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Baden LR, Rubin EJ. COVID-19—the search for effective therapy. N Engl J Med. 2020;382:1851–1852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Sheahan TP, Sims AC, Leist SR, et al. Comparative therapeutic efficacy of remdesivir and combination lopinavir, ritonavir, and interferon beta against MERS-CoV. Nat Commun. 2020;11:222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Cao B, Wang Y, Wen D, et al. A trial of Lopinavir–Ritonavir in adults hospitalized with severe COVID-19. N Engl J Med. 2020;382:1787–1799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Valk SJ, Piechotta V, Chai KL, et al. Convalescent plasma or hyperimmune immunoglobulin for people with COVID-19: rapid review. Cochrane Database Syst Rev. 2020;5:Cd013600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.McCreary EK, Pogue JM. Coronavirus disease 2019 treatment: a review of early and emerging options. Open Forum Infect Dis. 2020;7:ofaa105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Xu S, Li Y. Beware of the second wave of COVID-19. Lancet. 2020;395:1321–1322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Burton DR, Walker LM. Rational vaccine design in the time of COVID-19. Cell Host Microbe. 2020;27:695–698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Jenner E. Dr. Jenner, on the vaccine inoculation. Med Phys J. 1800;3:502–503. [PMC free article] [PubMed] [Google Scholar]
- 24.Nabel GJ. Designing tomorrow’s vaccines. N Engl J Med. 2013;368:551–560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Pronker ES, Weenen TC, Commandeur H, et al. Risk in vaccine research and development quantified. PLoS One. 2013;8:e57755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Clinical Trial-Related Terms. Grants and Funding. National Institute of Health (NIH)Available at https://grants.nih.gov/policy/clinical-trials/glossary-ct.htm. Accessed June 22, 2020. [Google Scholar]
- 27.Clinical Trials Phases. National Institute of Health (NIH). Available at https://www.clinicaltrials.gov/ct2/help/glossary/phase. Accessed June 22, 2020.
- 28.Clinical Research. Food and Drug Administration (FDA). Available at https://www.fda.gov/patients/drug-development-process/step-3-clinical-research. Accessed June 22, 2020.
- 29.Centers for Disease Control and Prevention (CDC). Principles of Vaccination. Available at: https://www.cdc.gov/vaccines/pubs/pinkbook/prinvac.html. Accessed July 12, 2020.
- 30.Abraham J. Passive antibody therapy in COVID-19. Nat Rev Immunol. 2020;20:401–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Sewell HF, Agius RM, Kendrick D, et al. Vaccines, convalescent plasma, and monoclonal antibodies for COVID-19. BMJ. 2020;370:m2722. [DOI] [PubMed] [Google Scholar]
- 32.Casadevall A, Joyner MJ, Pirofski LA. A randomized trial of convalescent plasma for COVID-19-potentially hopeful signals. JAMA. 2020;324:455–457. [DOI] [PubMed] [Google Scholar]
- 33.Li L, Zhang W, Hu Y, et al. Effect of convalescent plasma therapy on time to clinical improvement in patients with severe and life-threatening COVID-19: a JAMA. 2020;324:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Piechotta V, Chai KL, Valk SJ, et al. Convalescent plasma or hyperimmune immunoglobulin for people with COVID-19: a living systematic review. Cochrane Database Syst Rev. 2020;7:Cd013600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Clem AS. Fundamentals of vaccine immunology. J Glob Infect Dis. 2011;3:73–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Vaccine Types. Vaccines.gov; Available at https://www.vaccines.gov/basics/types. Accessed June 24, 2020. [Google Scholar]
- 37.Versteeg L, Almutairi MM, Hotez PJ, et al. Enlisting the mRNA vaccine platform to combat parasitic infections. Vaccines (Basel). 2019;7:122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Zhang C, Maruggi G, Shan H, et al. Advances in mRNA vaccines for infectious diseases. Front Immunol. 2019;10:594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Naran K, Nundalall T, Chetty S, et al. Principles of immunotherapy: implications for treatment strategies in cancer and infectious diseases. Front Microbiol. 2018;9:3158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Pandey SC, Pande V, Sati D, et al. Vaccination strategies to combat novel corona virus SARS-CoV-2. Life Sci. 2020;256:117956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Rauch S, Jasny E, Schmidt KE, et al. New vaccine technologies to combat outbreak situations. Front Immunol. 2018;9:1963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Lurie N, Saville M, Hatchett R, et al. Developing COVID-19 vaccines at pandemic speed. N Engl J Med. 2020;382:1969–1973. [DOI] [PubMed] [Google Scholar]
- 43.Mascola JR, Fauci AS. Novel vaccine technologies for the 21 century. Nat Rev Immunol. 2020;20:87–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Bouard D, Alazard-Dany D, Cosset FL. Viral vectors: from virology to transgene expression. Br J Pharmacol. 2009;157:153–165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Ramezanpour B, Haan I, Osterhaus A, et al. Vector-based genetically modified vaccines: exploiting Jenner’s legacy. Vaccine. 2016;34:6436–6448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Lee CS, Bishop ES, Zhang R, et al. Adenovirus-mediated gene delivery: potential applications for gene and cell-based therapies in the new era of personalized medicine. Genes Dis. 2017;4:43–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Lauer KB, Borrow R, Blanchard TJ. Multivalent and multipathogen viral vector vaccines. Clin Vaccine Immunol. 2017;24:e00298–e00316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Tan WG, Jin HT, West EE, et al. Comparative analysis of simian immunodeficiency virus gag-specific effector and memory CD8+ T cells induced by different adenovirus vectors. J Virol. 2013;87:1359–1372.23175355 [Google Scholar]
- 49.Liu MA. DNA vaccines: an historical perspective and view to the future. Immunol Rev. 2011;239:62–84. [DOI] [PubMed] [Google Scholar]
- 50.Pardi N, Hogan MJ, Porter FW, et al. mRNA vaccines – a new era in vaccinology. Nat Rev Drug Discov. 2018;17:261–279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Probst J, Weide B, Scheel B, et al. Spontaneous cellular uptake of exogenous messenger RNA in vivo is nucleic acid-specific, saturable and ion dependent. Gene Ther. 2007;14:1175–1180. [DOI] [PubMed] [Google Scholar]
- 52.Wang F, Kream RM, Stefano GB. An evidence based perspective on mRNA-SARS-CoV-2 vaccine development. Med Sci Monit. 2020;26:e924700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Reichmuth AM, Oberli MA, Jaklenec A, et al. mRNA vaccine delivery using lipid nanoparticles. Ther Deliv. 2016;7:319–334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Weiss TJ, Scheiblhofer R. Sandra. Gene Vaccines. 2012. [Google Scholar]
- 55.Liu MA. A comparison of plasmid DNA and mRNA as vaccine technologies. Vaccines (Basel). 2019;7:37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Wolff JA, Malone RW, Williams P, et al. Direct gene transfer into mouse muscle in vivo. Science. 1990;247(4949 Pt 1):1465–1468. [DOI] [PubMed] [Google Scholar]
- 57.Sahin U, Karikó K, Türeci Ö. mRNA-based therapeutics—developing a new class of drugs. Nat Rev Drug Discov. 2014;13:759–780. [DOI] [PubMed] [Google Scholar]
- 58.Leonhardt C, Schwake G, Stögbauer TR, et al. Single-cell mRNA transfection studies: delivery, kinetics and statistics by numbers. Nanomedicine. 2014;10:679–688. [DOI] [PubMed] [Google Scholar]
- 59.McCarthy M. DNA vaccination: a direct line to the immune system. Lancet. 1996;348:1232. [DOI] [PubMed] [Google Scholar]
- 60.Guimaraes PPG, Zhang R, Spektor R, et al. Ionizable lipid nanoparticles encapsulating barcoded mRNA for accelerated in vivo delivery screening. J Control Release. 2019;316:404–417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Kutzler MA, Weiner DB. DNA vaccines: ready for prime time? Nat Rev Genet. 2008;9:776–788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Wang H, Li X, Li T, et al. The genetic sequence, origin, and diagnosis of SARS-CoV-2. Eur J Clin Microbiol Infect Dis. 2020;39:1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Hoffmann M, Kleine-Weber H, Schroeder S, et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell. 2020;181:271–280.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Bouhaddou M, Memon D, Meyer B, et al. The global phosphorylation landscape of SARS-CoV-2 infection. Cell. 2020;182:685–712.e19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Zhou P, Yang XL, Wang XG, et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature. 2020;579:270–273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Walls AC, Park YJ, Tortorici MA, et al. Structure, function, and antigenicity of the SARS-CoV-2 spike glycoprotein. Cell. 2020;181:281–292.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.World Health Organization. Draft Landscape of COVID-19 Candidate Vaccines. Available at https://www.who.int/publications/m/item/draft-landscape-of-Covid-19-candidate-vaccines. Accessed June 9, 2020.
- 68.Chinese Clincial Trails Register. A Phase I/II Clinical Trial for Inactivated Novel Coronavirus (2019-CoV) Vaccine (Vero cells). Registration number: ChiCTR2000032459. Available at http://www.chictr.org.cn/showproj.aspx?proj=53003. Accessed June 9, 2020.
- 69.Chinese Clincial Trails Register. A Randomized, Double-blind, Placebo Parallel-controlled Phase I/II Clinical Trial for Inactivated Novel Coronavirus Pneumonia vaccine (Vero cells). Registration number: ChiCTR2000031809. Available at http://www.chictr.org.cn/showprojen.aspx?proj=52227. Accessed June 9, 2020.
- 70.U.S. National Library of Medicine. Safety and Immunogenicity Study of Inactivated Vaccine for Prevention of SARS-CoV-2 Infection(COVID-19). ClinicalTrials.gov Identifier: NCT04383574. Available at https://clinicaltrials.gov/ct2/show/NCT04383574?term=covid-19&cond=vaccine&cntry=CN&draw=2. Accessed June 9, 2020.
- 71.U.S. National Library of Medicine. Safety and Immunogenicity Study of an Inactivated SARS-CoV-2 Vaccine for Preventing Against COVID-19. ClinicalTrials.gov Identifier: NCT04412538. Available at https://clinicaltrials.gov/ct2/show/NCT04412538?term=vaccine&cond=covid-19&draw=2. Accessed June 9, 2020.
- 72.U.S. National Library of Medicine. Clinical Trial of Efficacy and Safety of Sinovac’s Adsorbed COVID-19 (Inactivated) Vaccine in Healthcare Professionals (PROFISCOV). ClinicalTrials.gov. Available at https://clinicaltrials.gov/ct2/show/NCT04456595?term=vaccine&cond=covid-19&draw=2&rank=1. Accessed July 12, 2020.
- 73.U.S. National Library of Medicine. Whole-Virion Inactivated SARS-CoV-2 Vaccine (BBV152) for COVID-19 in Healthy Volunteers (BBV152). Available at https://clinicaltrials.gov/ct2/show/NCT04471519?term=bharat&cond=covid-19&draw=2&rank=1. Accessed August 13, 2020.
- 74.U.S. National Library of Medicine. Evaluation of the Safety and Immunogenicity of a SARS-CoV-2 rS (COVID-19) Nanoparticle Vaccine with/without Matrix-M Adjuvant. ClinicalTrials.gov Identifier: NCT04368988. Available at https://clinicaltrials.gov/ct2/show/NCT04368988?term=vaccine&recrs=a&cond=covid-19&draw=2. Accessed June 9, 2020.
- 75.ClinicalTrials.gov. SCB-2019 as COVID-19 Vaccine. Available at https://clinicaltrials.gov/ct2/show/NCT04405908?term=clover&cond=covid-19&draw=2&rank=1. Accessed July 12, 2020.
- 76.U.S. National Library of Medicine. Phase I Clinical Study of Recombinant Novel Coronavirus Vaccine. Available at https://clinicaltrials.gov/ct2/show/NCT04445194?term=longcom&draw=2. Accessed July 12, 2020.
- 77.U.S. National Library of Medicine. Monovalent Recombinant COVID19 Vaccine (COVAX19). Available at https://clinicaltrials.gov/ct2/show/NCT04453852?term=vaccine&cond=covid-19&draw=5. Accessed July 12, 2020.
- 78.EU Clinical Trials Register. A Phase 2/3 Study to Determine the Efficacy, Safety and Immunogenicity of the Candidate Coronavirus Disease (COVID-19) vaccine ChAdOx1 nCoV-19. EudraCT number: 2020-001228-32. Available at https://www.clinicaltrialsregister.eu/ctr-search/trial/2020-001228-32/GB. Accessed June 9, 2020.
- 79.EU Clinical Trials Register. A Phase I/II Study to Determine Efficacy, Safety and Immunogenicity of the Candidate Coronavirus Disease (COVID-19) Vaccine ChAdOx1 nCoV-19 in UK Healthy Adult Volunteers. EudraCT number: 2020-001072-15. Available at https://www.clinicaltrialsregister.eu/ctr-search/trial/2020-001072-15/GB. Accessed June 9, 2020.
- 80.Chinese Clincial Trails Register. A Phase I Clinical Trial for Recombinant Novel Coronavirus (2019-COV) Vaccine (adenoviral vector). Registration number: ChiCTR2000030906. Available at http://www.chictr.org.cn/showprojen.aspx?proj=51154. Accessed June 9, 2020.
- 81.Chinese Clincial Trails Register. A Randomized, Double-blinded, Placebo-controlled Phase II Clinical Trial for Recombinant Novel Coronavirus (2019-nCOV) Vaccine (Adenovirus Vector). Registration number: ChiCTR2000031781. Available at http://www.chictr.org.cn/showprojen.aspx?proj=52006. Accessed June 9, 2020. [Google Scholar]
- 82.U.S. National Library of Medicine. An Open Study of the Safety, Tolerability and Immunogenicity of the Drug “Gam-COVID-Vac” Vaccine against COVID-19. ClinicalTrials.gov. Available at https://clinicaltrials.gov/ct2/show/NCT04436471?term=vaccine&cond=covid-19&draw=4. Accessed June 25, 2020.
- 83.ISRCTNRegistry. A phase III study to investigate a vaccine against COVID-19. Available at http://www.isrctn.com/ISRCTN89951424. Accessed June 25, 2020.
- 84.U.S. National Library of Medicine. A Study of Ad26.COV2.S in Adults (COVID-19). Available at https://clinicaltrials.gov/ct2/show/NCT04436276?term=NCT04436276&draw=2&rank=1. Accessed August 13, 2020.
- 85.U.S. National Library of Medicine. Dose-Confirmation Study to Evaluate the Safety, Reactogenicity, and Immunogenicity of mRNA-1273 COVID-19 Vaccine in Adults Aged 18 Years and Older. ClinicalTrials.gov Identifier: NCT04405076. Available at https://clinicaltrials.gov/ct2/show/NCT04405076?term=moderna&cond=covid-19&draw=2&rank=1. Accessed June 5, 2020.
- 86.U.S. National Library of Medicine. Safety and Immunogenicity Study of 2019-nCoV Vaccine (mRNA-1273) for Prophylaxis of SARS-CoV-2 Infection (COVID-19). ClinicalTrials.gov Identifier: NCT04283461. Available at https://clinicaltrials.gov/ct2/show/NCT04283461?term=vaccine&cond=covid-19&draw=2. Accessed June 5, 2020.
- 87.Safety, Tolerability and Immunogenicity of INO-4800 for COVID-19 in Healthy Volunteers. ClinicalTrials.gov Identifier: NCT04336410. Available at https://clinicaltrials.gov/ct2/show/NCT04336410?term=inovio&cond=covid-19&draw=2&rank=1. Accessed June 9, 2020.
- 88.Study to Describe the Safety, Tolerability, Immunogenicity, and Potential Efficacy of RNA Vaccine Candidates Against COVID-19 in Healthy Adults. ClinicalTrials.gov Identifier: NCT04368728. Available at https://clinicaltrials.gov/ct2/show/NCT04368728?term=vaccine&cond=covid-19&draw=3. Accessed June 5, 2020.
- 89.Clinical trial to assess the safety of a coronavirus vaccine in healthy men and women. Imperial College London; Available at http://www.isrctn.com/ISRCTN17072692. Accessed June 25, 2020. [Google Scholar]
- 90.A Study to Evaluate the Safety, Reactogenicity and Immunogenicity of Vaccine CVnCoV in Healthy Adults. Available at https://clinicaltrials.gov/ct2/show/NCT04449276?term=vaccine&cond=covid-19&draw=6. Accessed July 12, 2020.
- 91.A Phase I Clinical Trial to Evaluate the Safety, Tolerance and Preliminary Immunogenicity of Different Doses of a SARS-CoV-2 mRNA Vaccine in Population Aged 18–59 years and 60 years and above. Available at http://www.chictr.org.cn/showprojen.aspx?proj=55524. Accessed July 12, 2020.
- 92.Clinical Trials Registry – India. Novel Corona Virus-2019-nCov Vaccine by Intradermal Route in Healthy Subjects. Available at http://ctri.nic.in/Clinicaltrials/pmaindet2.php?trialid=45306&EncHid=&userName=vaccine2020.
- 93.Clinicaltrials.gov. Safety and Immunogenicity Study of GX-19, a COVID-19 Preventive DNA Vaccine in Healthy Adults. Available at https://clinicaltrials.gov/ct2/show/NCT04445389?term=vaccine&cond=covid-19&draw=3. Accessed July 12, 2020.
- 94.Osaka University/ AnGes/ Takara Bio. Available at https://www.clinicaltrials.jp/cti-user/trial/Show.jsp. Accessed July 12, 2020.
- 95.Zhu FC, Li YH, Guan XH, et al. Safety, tolerability, and immunogenicity of a recombinant adenovirus type-5 vectored COVID-19 vaccine: dose-escalation, open-label, non-randomised, first-in-human trial. Lancet. 2020;395:1845–1854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Humphreys IR, Sebastian S. Novel viral vectors in infectious diseases. Immunology. 2018;153:1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Fausther-Bovendo H, Kobinger GP. Pre-existing immunity against Ad vectors: humoral, cellular, and innate response, what’s important? Hum Vaccin Immunother. 2014;10:2875–2884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Zhu FC, Guan XH, Li YH, et al. Immunogenicity and safety of a recombinant adenovirus type-5-vectored COVID-19 vaccine in healthy adults aged 18 years or older: a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet. 2020;396:479–488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Bangari DS, Mittal SK. Development of nonhuman adenoviruses as vaccine vectors. Vaccine. 2006;24:849–862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Folegatti PM, Ewer KJ, Aley PK, et al. ; Oxford COVID Vaccine Trial Group. Safety and immunogenicity of the ChAdOx1 nCoV-19 vaccine against SARS-CoV-2: a preliminary report of a phase ½, single-blind, randomised controlled trial. Lancet. 2020;396:467–478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.COVID-19 Oxford Vaccine Trial. The Oxford Vaccine Centre COVID-19 Phase II/III Clinical Trial Explained. Available at https://covid19vaccinetrial.co.uk/phase-iiiii-trial-explained. Accessed June 23, 2020.
- 102.Jackson LA, Anderson EJ, Rouphael NG, et al. An mRNA vaccine against SARS-CoV-2-preliminary report. N Engl J Med. 2020:NEJMoa2022483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Kirkcaldy RD, King BA, Brooks JT. COVID-19 and postinfection immunity: limited evidence, many remaining questions. JAMA. 2020;323:2245–2246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Ota M. Will we see protection or reinfection in COVID-19? Nat Rev Immunol. 2020;20:351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Roy S. COVID-19 reinfection: myth or truth? SN Compr Clin Med. 2020;29:1–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Lan L, Xu D, Ye G, et al. Positive RT-PCR test results in patients recovered from COVID-19. JAMA. 2020;323:1502–1503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Bentivegna E, Sentimentale A, Luciani M, et al. New IgM seroconversion and positive RT-PCR test after exposure to the virus in recovered COVID-19 patient. J Med Virol. 2020:10.1002/jmv.26160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Mo H, Zeng G, Ren X, et al. Longitudinal profile of antibodies against SARS-coronavirus in SARS patients and their clinical significance. Respirology. 2006;11:49–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Payne DC, Iblan I, Rha B, et al. Persistence of antibodies against Middle East respiratory syndrome coronavirus. Emerg Infect Dis. 2016;22:1824–1826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Batisse D, Benech N, Botelho-Nevers E, et al. Clinical recurrences of COVID-19 symptoms after recovery: viral relapse, reinfection or inflammatory rebound? J infect. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Chandrashekar A, Liu J, Martinot AJ, et al. SARS-CoV-2 infection protects against rechallenge in rhesus macaques. Science. 2020;369:812–817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Long QX, Liu BZ, Deng HJ, et al. Antibody responses to SARS-CoV-2 in patients with COVID-19. Nat Med. 2020;26:845–848. [DOI] [PubMed] [Google Scholar]
- 113.Watson J, Whiting PF, Brush JE. Interpreting a COVID-19 test result. BMJ. 2020;369:m1808. [DOI] [PubMed] [Google Scholar]
- 114.Shen C, Wang Z, Zhao F, et al. Treatment of 5 critically ill patients with COVID-19 with convalescent plasma. JAMA. 2020;323:1582–1589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Xing Y, Mo P, Xiao Y, et al. Post-discharge surveillance and positive virus detection in two medical staff recovered from coronavirus disease 2019 (COVID-19), China, January to February 2020. Euro Surveill. 2020;25:2000191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Xiao AT, Tong YX, Zhang S. False-negative of RT-PCR and prolonged nucleic acid conversion in COVID-19: rather than recurrence. J Med Virol. 2020:10.1002/jmv.25855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Kang H, Wang Y, Tong Z, et al. Retest positive for SARS-CoV-2 RNA of “recovered” patients with COVID-19: persistence, sampling issues, or re-infection? J Med Virol. 2020:10.1002/jmv.26114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.To KK, Tsang OT, Leung WS, et al. Temporal profiles of viral load in posterior oropharyngeal saliva samples and serum antibody responses during infection by SARS-CoV-2: an observational cohort study. Lancet Infect Dis. 2020;20:565–574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Wölfel R, Corman VM, Guggemos W, et al. Virological assessment of hospitalized patients with COVID-2019. Nature. 2020;581:465–469. [DOI] [PubMed] [Google Scholar]
- 120.Ortega JT, Serrano ML, Pujol FH, et al. Role of changes in SARS-CoV-2 spike protein in the interaction with the human ACE2 receptor: an in silico analysis. Excli J. 2020;19:410–417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Favre D, Provost N, Blouin V, et al. Immediate and long-term safety of recombinant adeno-associated virus injection into the nonhuman primate muscle. Mol Ther. 2001;4:559–566. [DOI] [PubMed] [Google Scholar]
- 122.Armengol G, Ruiz LM, Orduz S. The injection of plasmid DNA in mouse muscle results in lifelong persistence of DNA, gene expression, and humoral response. Mol Biotechnol. 2004;27:109–118. [DOI] [PubMed] [Google Scholar]
- 123.Wang Z, Troilo PJ, Wang X, et al. Detection of integration of plasmid DNA into host genomic DNA following intramuscular injection and electroporation. Gene Ther. 2004;11:711–721. [DOI] [PubMed] [Google Scholar]
- 124.Ura T, Okuda K, Shimada M. Developments in viral vector-based vaccines. Vaccines (Basel). 2014;2:624–641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Fló J, Tisminetzky S, Baralle F. Modulation of the immune response to DNA vaccine by co-delivery of costimulatory molecules. Immunology. 2000;100:259–267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Alberer M, Gnad-Vogt U, Hong HS, et al. Safety and immunogenicity of a mRNA rabies vaccine in healthy adults: an open-label, non-randomised, prospective, first-in-human phase 1 clinical trial. Lancet. 2017;390:1511–1520. [DOI] [PubMed] [Google Scholar]
- 127.Schlake T, Thess A, Thran M, et al. mRNA as novel technology for passive immunotherapy. Cell Mol Life Sci. 2019;76:301–328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Lithander FE, Neumann S, Tenison E, et al. COVID-19 in older people: a rapid clinical review. Age Ageing. 2020;49:501–515. [DOI] [PMC free article] [PubMed] [Google Scholar]